Effects of Postnatal Glucocorticoids on Brain Structure in Preterm Infants, A Scoping Review ============================================================================================ * Isabella Robles * Margarita Alethea Eidsness * Katherine E. Travis * Heidi M Feldman * Sarah E. Dubner ## ABSTRACT Glucocorticoids (GC) are used in neonatal intensive care units to prevent or reduce the severity of chronic lung disease in preterm infants and have been implicated in impaired neurodevelopment. Our objective was to identify what is known about the effects of postnatal GC treatment in human preterm infants on structural brain development and to identify gaps in the literature. Following Arksey and O’Malley’s scoping review methodological framework, we searched scientific literature databases for original research on human preterm infants, postnatal GCs, and brain structure. 11 studies assessed the effects of GCs on structural brain outcomes. 56 studies reported brain injury, but not structure. Dexamethasone was consistently associated with decreased total and regional brain volumes, including cerebellar volumes. Hydrocortisone was often, but not always associated with absence of brain volume differences. No studies examined the impact of inhaled GC on brain structure. Additional research on the effects of neonatal GCs after preterm birth on a variety of structural brain measures is required for understanding contributions to neurodevelopment and informing practice guidelines. **HIGHLIGHTS** * Few studies examine postnatal glucocorticoid effects on preterm human brain structure * Dexamethasone use in preterm infants is associated reduced overall brain volumes * Dexamethasone use in preterm infants is associated with reduced cerebellar volumes * Hydrocortisone use in preterm infants is not consistently correlated with adverse brain effects * Studies of glucocorticoid use in preterm infants should assess brain structures KEYWORDS * glucocorticoids * prematurity * brain * imaging * human ## 1.0 INTRODUCTION Glucocorticoids (GC) are a class of steroid hormones that may be produced endogenously by the adrenal gland or provided exogenously as an anti-inflammatory or immunosuppressant medication. Exogenous GC are highly effective as anti-inflammatory medications, but are associated with multiple complications, especially after prolonged use. Exogenous GC are graded in terms of potency; dexamethasone is a potent GC whereas hydrocortisone is a less potent GC. Infants born preterm (PT), particularly those born more than 2 months before their due date, labelled very and extremely preterm, often require support for basic bodily functions, such as mechanical ventilation for respiratory illness. These PT children are at high risk for multiple inflammatory complications during their neonatal hospitalization, including a severe inflammatory chronic lung disease, called bronchopulmonary dysplasia (BPD). They are also at risk for long-term neurodevelopmental disorders, particularly among those children with inflammatory conditions, such as BPD (Gallini et al., 2021; Patra et al., 2017). The use of exogenous GC for infants born preterm (PT) in the neonatal intensive care unit has been a controversial topic, much debated in recent decades. GCs are commonly administered to PT infants to reduce the incidence and severity of BPD, which, in turn, is a major risk factor for mortality and neurodevelopmental disability in children born PT. The overall aim of this scoping review is to review the existing literature in human infants to determine what is known about the impact of exogenous GC, administered in the neonatal intensive care unit to prevent or reduce the severity of BPD, in relation to early brain structural development. Despite widespread use, multiple investigations have questioned whether administration of GC is appropriate for preterm infants for several important reasons: (1) studies have failed to show a clear and consistent benefit of the medication in decreasing rates or severity of BPD (Cummings et al., 2022), (2) some studies have shown poor neurodevelopmental outcomes in GC-treated preterm infants, especially after administration of dexamethasone (Cummings et al., 2022), and (3) animal studies have found adverse effect of GCs on brain structures (Huang et al., 1999). It is generally assumed that brain structural metrics are associated both with neonatal clinical experience (Volpe, 2009) and with neurodevelopmental outcomes (Ment et al., 2009). Thus, we embarked on this review. We anticipated variation in the types of GCs used and in the methods of neuroimaging used to evaluate brain structure, both limiting our ability to conduct a systematic review and meta-analysis. We therefore conceptualized this review as a scoping review to synthesize what is currently known in the field, identify gaps in the literature, and direct future research questions. ### 1.1 Glucocorticoid use in preterm infants Systematic use of postnatal exogenous GC in PT infants for prevention of BPD followed a 1994 publication of recommendations for prenatal GC administration to reduce the likelihood of PT birth in women in PT labor (Noguchi, 2014). Subsequently, in 2002, mounting evidence of neuromotor and cognitive impairment and impaired cerebellar growth led the American Academy of Pediatrics (AAP) to recommend against the use of postnatal GC (Committee on Fetus and Newborn, 2002). However, since 2010 and the publication of several studies of lower dose and lower potency GCs, postnatal GC use has again increased. The most recent recommendations, published in 2022, recommended against high dose dexamethasone (Cummings et al., 2022). No recommendation was made regarding routine use of low potency steroids. Shared decision making between clinicians and parents was recommended when deciding whether to use steroids. In this state of clinical equipoise, additional information about steroid effects on brain structure may help clinicians and families choose a treatment path. ### 1.2 Outcomes after post-natal glucocorticoid administration in preterm infants Studies of long-term neurodevelopmental outcomes after GC administration in the neonatal period have been inconclusive regarding benefits and risks. Early studies were generally limited by small sample sizes, underpowered to describe effects of GCs on neurodevelopment and particularly to consider differences across different GCs, such as dexamethasone and hydrocortisone (Doyle, 2021). A recent large multicenter trial of over 800 infants failed to show benefit in decreasing BPD or improving survival without moderate to severe disability (Watterberg et al., 2022). The accompanying commentary suggested that research should adjust the dependent variables of interest and consider the role of GC in the amelioration of severe respiratory disease rather than in survival without BPD, as the primary measure of the utility of GC use (Greenough, 2022). To overcome the limitations of individual studies, multiple systematic reviews have considered the effects of GC administration and have made recommendations for GC administration in preterm infants (Chang, 2014; Doyle et al., 2021a, 2021a; Noguchi, 2014; Onland et al., 2017; Shah et al., 2017). These reviews focused on several issues: clinical outcomes, including prevalence of BPD and later neurodevelopmental outcomes, including neurodevelopmental disability; the optimal administration plan for steroids; and the appropriate dose to maximize effectiveness without incurring accompanying adverse effects. All these reviews concluded that further research on long-term effects needs to be done. They recommended minimal use of postnatal glucocorticoids, restricting treatment only to ventilator-dependent infants, and limiting dose and duration. A review specifically of early GC administration before 7 days of life concluded that the benefits of GCs (e.g. shorter ventilation time, decreases in diagnosed BPD), may not outweigh the known risks (e.g. gastrointenstinal bleeding, cerebral palsy, hyperglycemia) (Doyle et al., 2021b). As a result, the review recommended discontinuing the practice of early dexamethasone administration. A separate review specifically of steroids started after day of life 7 found that the risk of death and BPD was reduced without increased rates of cerebral palsy; however, the long term effects had not been well studied (Doyle et al., 2021a). This review demonstrated the importance of timing of GC administration. Concerning results were found in a review of specific steroid protocols (Onland et al., 2017). This review noted increased risk of BPD and adverse neurodevelopmental outcomes in children getting higher versus lower doses of GCs and no difference in outcomes for early versus late administration. The studies however had important methodological weaknesses and the review could not conclude with an optimal dose regimen recommendation. The reviews to date have not fully considered the effects of postnatal GCs on developing brain structures, even though alternations in brain structure may be a major contribution to adverse neurodevelopmental outcomes (Volpe, 2009). It remains critically important to understand any impacts because changes may relate to later neurodevelopmental outcomes. Indeed, structural brain findings may provide a proximal indicator, available to clinicians while children born preterm are still hospitalized, of the neurobiological effects of GCs, to inform clinical research and practice. ### 1.3 Glucocorticoid effects on brain development Several lines of evidence support consideration of the effects of GC on brain development in PT infants. GC enter the brain via simple diffusion across the blood brain barrier. Within the brain, GC affect brain development via intracellular glucocorticoid and mineralocorticoid receptors. Glucocorticoid receptor binding causes both inhibition and enhancement of gene transcription (De Kloet et al., 1998; Nishi and Kawata, 2007). Glucocorticoid receptor activation may suppress synaptic plasticity and inhibit neuronal development. Glucocorticoid receptors are widely expressed in the glia and neurons of the cerebrum and cerebellum (Bohn et al., 1994). Mineralocorticoid receptor activation meanwhile may aid in synapse plasticity and neuronal survival (Joëls, 2007; Johnston et al., 2009; McEwen, 1994; McEwen et al., 1992). Mineralocorticoid receptors are primarily found in the hippocampus and limbic structures (Joëls, 2001). Age and sex affect distribution; prefrontal glucocorticoid receptor density increases in childhood and expression in the hippocampus is higher in women than men. Importantly, dexamethasone primarily binds glucocorticoid receptors, while hydrocortisone additionally binds mineralocorticoid receptor (Reul et al., 2000), suggesting that the impact on brain structure may vary as a function of which GC was administered. The distinctive pharmacokinetic and pharmacodynamic profiles of exogenous GC during childhood may result in increased exposure of central nervous system tissue to exogenous glucocorticoids in children as compared to adults (Damsted et al., 2011). Preterm infants may see even higher GC exposure in the brain than older infants and children because of their permeable blood-brain barrier, reduced hepatic metabolism and renal excretion, and lower protein binding capacity. Animal studies provide additional evidence of potential effects of GC on infant brains. Studies in neonatal rats have shown decreased neurogenesis, impaired long term potentiation with dexamethasone, but not hydrocortisone. (Chang, 2014; Cotterrell et al., 1972; Howard and Granoff, 1968; Huang et al., 1999). These neural changes occurred along with behavioral abnormalities including delayed acquisition of motor skills and impaired emotional control. Early treatment in rats (postnatal days 2-8) have shown decreased hippocampal glucocorticoid receptor number at pubertal age (postnatal day 40), suggesting lasting impacts on the regulation of cellular development (Zoli et al., 1990). Adverse effects on CNS myelination have also been reported (Gumbinas et al., 1973). The cerebellum appears to be especially sensitive to GC exposure. Postnatal GC impair rat cerebellar development by decreasing cellular proliferation (Noguchi, 2014). Cerebellar development is coordinated in a time sensitive manner through proliferation of the extragranular layer, followed by a programmed, endogenous GC-induced apoptosis of external granular layer cells. This process, when induced too early by exogenous GC-administration, leads to impaired cerebellar development. Similarly, studies of human infants demonstrated decreased cerebellar volume after dexamethasone administration (Parikh NA et al., 2007; Tam et al., 2011). Evidence of hydrocortisone effects on cerebellum were mixed, compared with dexamethasone (Noguchi, 2014). ### 1.4 Rationale for review of post-natal glucocorticoids on brain structures Given the diversity of the studies and multifactorial contributors to neurodevelopmental outcomes, we sought to review the literature on a more proximal and objective measure, structural brain metrics, to inform our understanding of the effects of glucocorticoid administration on preterm brain development. We know that PT infants show a particular pattern of brain injury and dysmaturity due to complex interplay of developmental stage and medical intervention (Volpe, 2009). These brain changes are now detectable using advanced imaging techniques and the differences are seen in neurodevelopmental outcomes (Feldman et al., 2010). Several reviews have looked at imaging studies and GC as part of an overall review of steroid effects on the neonatal brain, the most recent of which are nearly 10 years old (Baud, 2004; Chang, 2014; Favrais et al., 2014; Noguchi, 2014; Rademaker and de Vries, 2009). Given the increase in imaging studies over the past decade, our aim was to review the current literature. Because of the diversity of GC types, doses, timing, duration, and the diversity of brain outcomes reported, a scoping review of the literature was most appropriate. The primary objective of this scoping review is to identify what is known about the effects of GC treatment on brain structural development in preterm human infants with the goal of identifying gaps in the literature and potential intermediate biomarker candidates for neurodevelopmental outcomes. With this review, we hope to assess the scope of current research on the topic and identify future directions for investigations of steroid administration and structural brain changes. ## 2.0 METHODS We followed the Arksey and O’Malley methodological framework for scoping reviews to ensure an orderly approach to mapping the existing evidence on what is known broadly about this topic and to identify gaps in the literature (Arksey and O’Malley, 2005). The stages include: (1) identifying the research question; (2) identifying relevant studies; (3) selecting the studies; (4) charting the data; and (5) collating, summarizing, and reporting the results. A search query was designed to yield articles that included 3 key topics: postnatal steroids, prematurity, and brain structure. We used a combination of MeSH terms and text word search of titles and abstracts. Our full search terms are shown in the **Supplement**. Using this strategy, we collected articles from [ClinicalTrials.gov](http://ClinicalTrials.gov) and PubMed published between January 1, 1990 and September 16, 2021. We also included references from recent Cochrane systematic reviews on postnatal glucocorticoid administration for preterm infants (Doyle et al., 2021a, 2021b; Onland et al., 2017). The inclusion and exclusion criteria are shown in **Table 1**. We defined a structural brain outcome as description or measurement of one or more anatomical features, assessed using one or more neuroimaging modalities. For example, MRI was commonly used to report on overall and regional brain volumes or image slice area. Studies that reported on structural brain outcomes after postnatal GC administration to preterm children were our primary interest in this review. We differentiated the anatomical descriptions and measurements from general descriptions, such as intraventricular, brain parenchymal, and/or cerebellar hemorrhages or injuries, which may or may not lead to variation in brain anatomy. These findings are frequently used to characterize preterm study populations and are not necessarily treated as an outcome of interest. We defined these outcomes as “brain injury”, reviewed them as a set, and, to provide a comprehensive view of the literature, included these references in the **Supplement**. Studies that included postnatal GC administration as a covariate or as a possible risk factor moderating relations between preterm birth and brain structure, but did not include information on GC type, dose, or administration were reviewed and also included in the **Supplement**. We excluded Near Infrared Spectroscopy (NIRS) and Doppler imaging modalities because they are considered functional modalities. View this table: [Table 1.](http://medrxiv.org/content/early/2022/11/11/2022.11.09.22282133/T1) Table 1. Inclusion and exclusion criteria for title and abstract review References were imported into Covidence (“Covidence systematic review software,” n.d.) and were screened by 3 independent reviewers (IR, AE, SD) at each step: title and abstract screening, full-text review, and data extraction. For title and abstract screening, references were excluded only if it obviously met exclusion criteria, such as being an animal study. For full-text review, articles were included if any structural brain outcome was reported, regardless of it being a primary or secondary outcome. The senior author (SD) reviewed and resolved conflicts when consensus through discussion was not reached. We extracted the following data from relevant studies: study design, aim of study, trial name, cohort size, steroid administration (type, dose, route, duration), imaging modality, primary and secondary outcomes, and brain outcome. For studies in the supplement, the reported outcomes were not structural brain outcomes. For these studies, we extracted the relevant brain data that were reported as a characteristic of the study population following steroid exposure. ## 3.0 RESULTS Using the criteria described in the methods, we identified 11 papers for inclusion in this review. A consort diagram is shown in the **Figure. Table 2** includes the studies that specifically examined structural brain outcomes as a result of GC exposure. An additional 62 studies were either glucocorticoid intervention trials that reported brain injury outcome (e.g. intraventricular hemorrhage, IVH) as a characteristic of the study population (n= 50), or were observational studies of brain structural outcomes that examined postnatal GC as a potential contributing or risk factor (n=12) and provided little or no information on glucocorticoid type or administration. These studies are summarized in the **Supplement.** ![FIGURE.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/11/11/2022.11.09.22282133/F1.medium.gif) [FIGURE.](http://medrxiv.org/content/early/2022/11/11/2022.11.09.22282133/F1) FIGURE. Consort diagram of studies screened and extracted. View this table: [Table 2.](http://medrxiv.org/content/early/2022/11/11/2022.11.09.22282133/T2) Table 2. Studies of glucocorticoids and brain structure in preterm born infants ### 3.1 DEXAMETHASONE Three studies reported on the association between dexamethasone exposure and a range of brain structural outcomes across a range of ages at the time of imaging. An early 3-D quantitative volumetric MRI study that imaged the participants at 38-41 weeks post conceptual age compared infants treated with dexamethasone to infants who were not exposed to GC and to term infants (Murphy BP et al., 2001). The dexamethasone-treated group was born at an earlier gestational age, had a longer length of stay, had higher clinical risk index for babies (CRIB) score in the first 12 hours of life. Cerebral cortical gray matter volume was reduced in the dexamethasone-exposed group compared with the non-exposed group, after adjusting for gestational age at birth and CRIB score (mean difference 65.3, 95% confidence interval: 4.3 to 114.58). There were no significant cerebral cortical gray matter volume differences between non-exposed preterm infants and control. Total cerebral tissue volume was reduced by 22% in the dexamethasone treated versus non-treated preterm groups. After adjustment for covariates, the volume remained lower, but was no longer statistically significant. No differences were observed in the subcortical gray matter volumes (basal ganglia and thalami). Myelinated white matter volume differed between preterm and full term groups, but was not different between dexamethasone exposed and non-exposed preterm infants. Unmyelinated white matter did not differ between groups. Parikh and colleagues (Parikh NA et al., 2007) examined a later sample with lower overall dexamethasone doses, adjusting for multiple potential confounders (PMA at scan, dexamethasone treatment, birthweight, presence of BPD). They obtained coronal T2 weighted MRI at a mean post menstrual age of 39.4 weeks. The authors reported smaller total brain volumes (9.5%), cerebellar volumes (19.7%) and subcortical gray matter volumes (20.6%) in the dexamethasone group. Total gray matter volume was also lower in the dexamethasone group but did not meet statistical significance. There was no dose response relationship between dexamethasone dose and total brain volume and cortical tissue volume. To explain the smaller reductions seen compared with Murphy et al. 2001, they pointed out that they used lower cumulative doses, later in life, to higher risk ELBW infants, enrolled an unselected cohort, performed masked evaluations, had a larger sample size, and adjusted for more potential confounders. A third study obtained T1 weighted MRI at age 18 years in a cohort of individuals born preterm who had received NICU administered dexamethasone. In this cohort, GC were given at the discretion of the treating clinicians after the first week of life. The dexamethasone group had a lower gestational age and birthweight and higher rates of BPD and cerebral palsy than those who did not receive postnatal dexamethasone. Overall they found a 3.6% (95% CI −7.0%, −0.3%) smaller total brain tissue volume at age 18, that was not significantly different between groups after adjustment for covariates. Volumes of total and most regional areas of cortical white matter, thalamus, and basal ganglia nuclei were smaller in the dexamethasone group compared with the no-dexamethasone group after adjustment for covariates. Unlike the studies reviewed here which looked at scans from younger ages, there was no statistically significant difference in cortical gray matter, hippocampus, amygdala, and cerebellar volumes. Within white matter, on the other hand, the reduction in brain parcel volumes in the dexamethasone group was found in most regions except the medial temporal region. The authors hypothesized that the absence of cerebellar volume differences may have been due to catch up cerebellar growth while the presence of white matter volume reductions found in their study, which had not been reported previously, may reflect a selective vulnerability of particular brain tissues and altered developmental trajectory that is not detected until adolescence. ### 3.2 HYDROCORTISONE Eight studies directly assessed the association between hydrocortisone and brain structure. In 2005, Lodygensky et. al obtained 3D quantitative volumetric MRI in 8 year old children who had been born preterm or full term (Lodygensky et al., 2005). A portion of the PT children had received hydrocortisone due to respiratory illness severity. No differences in 5 measures of brain volumes were seen between PT children who did or did not receive neonatal hydrocortisone. This study reports that it is the first directly to address the effect of postnatal corticosteroid treatment on hippocampal volumes in humans. The authors hypothesized that the combination of the decreased potency and shorter half-life of hydrocortisone compared with dexamethasone and hydrocortisone’s preferential binding of mineralocorticoid receptors versus dexamethasone’s binding of glucocorticoid receptors is protective. A larger, overlapping study of the 8 year old children with the same 3D quantitative volumetric MRI as Lodygensky study (Lodygensky et al., 2005) classified MRI findings as normal, mildly abnormal, or severely abnormal and calculated corpus callosum area on a midsagittal T2 slice. There were no differences in presence of brain lesions on MRI between PT children who were treated with hydrocortisone and those who were not after adjusting for a propensity score representing the likelihood of requiring hydrocortisone. Mean midsagittal corpus callosum area was smaller in the hydrocortisone treated group compared with the non-treated group, but the difference was not significant after adjustment for the propensity score. No other brain differences were identified. This investigation represented the largest group from their sample. In addition to the potency and half-life of hydrocortisone vs dexamethasone, the authors pointed to the use of sodium bisulphite as a preservative in dexamethasone as a possible cause of decreased neuronal cell line viability based on in vitro and rodent studies. They discussed the timing of glucocorticoid administration later in neonatal life as potentially being beyond a particular window of developmental vulnerability. Two studies reported on overlapping samples of PT infants with 3D volumetric MRI at term equivalent age (Benders et al., 2009; Kersbergen et al., 2013). Benders and colleagues performed a prospective study comparing infants who received a two-week course of hydrocortisone at 1 week of life or later with age, gender, and respiratory status matched controls from a second institution using the same MRI scanner and the same imaging protocol. Adjusted analyses showed no differences between groups for individual or summed volumes for the intracranial cavity, CSF, cortical gray matter, subcortical gray matter, unmyelinated white matter, and myelinated white matter. In the second investigation of an expanded cohort (Kersbergen et al., 2013) there was no difference in total brain tissue volume between treated and untreated controls after adjustment for covariates. Only post menstrual age at scan, birthweight, and gestational age at birth were associated with total brain tissue volume. This was the same whether hydrocortisone was treated as a continuous variable or high vs low dose. They also looked at cerebellar volumes and found that while PMA at scan, birth weight z score, and grade 3 IVH influenced cerebellar volumes, hydrocortisone did not. In a pilot randomized controlled trial, Parikh and colleagues obtained axial PD/T2 volumetric MRI scans at 38 weeks post menstrual age in infants randomized to hydrocortisone or placebo with the specific objective of examining a structural brain outcome (Parikh et al., 2013). Total brain tissue volume was the primary outcome. Secondary brain outcomes included individual tissue volumes of cortical gray matter, cerebral white matter, cerebrospinal fluid, subcortical gray matter and substructure volumes of included nuclei, cerebellum, hippocampi, amygdalae, corpus callosum, and brain stem. The study was powered to detect a 2-week growth difference in brain size. In bivariate and adjusted analyses, total tissue volume was not different between the groups. Secondary brain outcomes of regional volumes were not different between the groups. The authors concluded that there were neither short term benefits or harms from low dose hydrocortisone after the first week of life. Allison et al. (Alison M et al., 2020) conducted a large predefined secondary analysis of T1, T2, T2 GE MRI at term equivalent age of patients enrolled in the PREMILOC trial of early hydrocortisone. A standardized scoring system was used to evaluate cerebral WM, cortical and deep GM and cerebellar abnormalities. (Kidokoro et al., 2013) A number of quantitative biometric values (sizes and areas of various structures) were obtained. The primary endpoint was the occurrence of cerebral white matter abnormalities. There were no differences in the cumulative distribution of white matter scores between the treatment and control groups. Fewer infants born at 24-25 weeks in the hydrocortisone group developed dilated lateral ventricles compared with the placebo group. The 24-25 week hydrocortisone group also had smaller lateral ventricles than the placebo group. There were no differences in any of the cortical and basal ganglia gray matter, nor cerebellar injury between the groups. There was no difference in the distribution of the relevant scores between groups. There was a statistically significant association between overall brain injury score cumulative distribution between the groups, however hydrocortisone was no longer significantly associated with white matter damage or overall moderate to severe brain damage after adjustment for risk factors and gestational age. Rousseau (Rousseau et al., 2021) obtained axial T2 MRI and aimed to explore postnatal brain growth in extremely preterm born infants requiring postnatal steroids. They examined brain tissue area at term equivalent age and head circumference growth by 12 months of age. This report included children receiving hydrocortisone for any indication (e.g., refractory hypotension, prolonged respiratory insufficiency). Semiautomatic tissue segmentation was performed and areas of each tissue were measured on an axial slice (not a 3D volumetric measurement). The study was powered to see an 8% reduction of brain volumes at term equivalent age. Despite matching, the hydrocortisone group was lower weight at birth and had more medical complications. Adjusting for postmenstrual age at MRI scan, there was a significant reduction in the intracranial cavity, the basal ganglia and thalamus area at term equivalent age. There was no correlation between basal ganglia and thalamus (BGT) area and hydrocortisone dose. Multiple regression showed that duration of mechanical ventilation was the only significant independent variable associated with BGT (not with hydrocortisone treatment). ### 3.3 HYDROCORTISONE AND DEXAMETHASONE Tam and colleagues published a study focused on steroid effects on the cerebrum and cerebellum that examined both hydrocortisone and dexamethasone (Tam et al., 2011). They obtained T2-weighted and T1 3-D volumetric scans at two sites at two time points – as soon after birth as the infant was able and again at term post menstrual age. Adjusting for multiple covariates, there was a decrease in cerebellar growth associated with glucocorticoid exposure resulting in 1.88 cm3 smaller cerebellum at 40 weeks post menstrual age after hydrocortisone and 2.31 cm3 smaller cerebellum associated with dexamethasone (8% and 10% smaller cerebellar volumes, respectively). They were unable to determine whether there was a dose dependent effect of glucocorticoids on cerebellar growth. Cerebellar volume was not associated with the number of days since the last steroid administration, indicating that the volume changes were not immediately reversible effects. Clinical factors were also associated with decreased cerebellar volume including intubation duration, hypotension, patent ductus arteriosus, but postnatal dexamethasone, postnatal hydrocortisone, and severe intraventricular hemorrhage were associated with the largest decreases in cerebellar volume. The same analyses were performed for cerebral volumes at term. Cerebral volume at term was not associated with antenatal betamethasone exposure or with postnatal hydrocortisone or dexamethasone exposure. ### 3.4 INHALED GLUCOCORTICOIDS We identified no studies examining association between inhaled corticosteroids in preterm infants and structural brain outcomes. ## 4.0 DISCUSSION In this scoping review we identified 11 papers related to associations or effects between postnatal GC and structural or anatomical brain metrics in humans born PT. Dexamethasone was consistently associated with unfavorable brain differences. Radiologist scoring rubrics and quantitative total and regional brain areas or volumes of varied structures were reported. The results showed that for dexamethasone, exposure to dexamethasone was associated with reduction in volumes across multiple brain regions, including the cerebellum, and metrics. However, for hydrocortisone, only one study has reported that exposure to medication was associated with volumetric differences and these localized to the cerebellum. Overall cohort sizes varied from small to large. The range of treatment dose, duration, and age at initiation varied widely. ### 4.1 STUDY TIMING While most studies examined imaging obtained during the initial neonatal course, there were several reports on a cohort of 8-year-old children born preterm and one as late as 18 years of age. All of these time frames are important and relevant to understanding the potential long-term outcome of GC given during a sensitive developmental window. The case for using imaging during the neonatal hospital course is strong. Infant MRI metrics may serve as proximal biomarkers associated with later developmental outcomes. Having a predictive biomarker in the neonatal period can avoid the confounding of later post-NICU experience in understanding the effects of GC on brain. Future studies should assess advanced MRI metrics as mediators of GC effects on later neurodevelopmental outcomes. Evaluation at later ages is also important to understanding neurodevelopment. The preterm newborn brain undergoes rapid development. Glucocorticoids in the neonatal period act on a changing substrate. Effects may not be evident until the brain structure is more mature. Furthermore, the timing of glucocorticoid administration during the neonatal period could produce different effects on brain structure and development. ### 4.2 MEASUREMENT CHOICE In the clinical setting, a measure should be objective, easily obtained, relevant to the exposure and the later outcome it relates to. Because of the differing GR and MR receptor distribution in the brain, examining subregions or certain types of brain matter (e.g. white vs gray matter; cerebral vs cerebellar), may better identify effects than a whole brain approach. Although not routinely implemented in the clinical setting, no study has yet examined effects of GC using advanced microstructural metrics, for instance diffusion metrics (Feldman et al., 2010) or quantitative microstructural metrics (Mancini et al., 2020). These metrics may get closer to the underlying neurobiology and therefore may detect GC effects that cannot be seen on the more macroscopic volumetric scale that has been reported thus far. Using these quantitative metrics may shed light on contradictory results from the macroscopic volumetric measures. ### 4.3 CHALLENGES IN CONDUCTING AND COMPARING STUDIES This review highlights the difficulty of conducting and comparing randomized trials of steroids in preterm infants. Some studies were found not to be feasible due to difficulty with patient recruitment. Studies were stopped, and dosing changed midway. Most of the studies reporting on brain structure were small or of modest size. Changes in clinical practice over time make comparison across studies challenging. The larger studies often had a range of GC doses and regimens. While multicenter RCTs should incorporate brain metrics into their study design, we should also consider how prospective or retrospective cohorts can account for heterogeneity in treatment and sample size limitations to leverage the power of modern electronic health records in asking clinical questions such as those relating to GC use. Rousseau (Rousseau C et al., 2021) for instance investigated correlations between dose and brain area, while Kersbergen (Kersbergen et al., 2013) looked at both continuous and high/low measures. Tam et. al. employed several strategies for analyzing cohort data (Tam et al., 2011). The multi-site study was able to collect a large sample. They were the only study to examine brain imaging longitudinally, strengthening the causal argument. They examined regional brain volumes, and they attempted to assess dose response. Their results have not been reproduced, although it is difficult to directly compare studies because of the analytic complexities discussed here. Collaboration and continued research with more current datasets will be important to understanding the safety and effectiveness of GC treatments in preterm infants. ### 5.0 CONCLUSIONS GC effects on brain are of interest to a wide audience of researchers across the lifespan and across many clinical conditions. Despite the availability of clinical and research advanced imaging modalities, relatively few human studies have directly assessed the effect of this intervention on brain structural development. This review highlights the need for additional research on neonatal GC and their potential effects on brain development. As GC use becomes more targeted and ongoing research aims to identify optimal populations and treatment regimens, incorporating information about both brain structure in the infant period and in later childhood can provide researchers and clinicians with a better understanding of the downstream effects of this important treatment on neurodevelopment. ## Supporting information PRISMA_Checklist [[supplements/282133_file03.docx]](pending:yes) ## Data Availability All data produced in the present study are available upon reasonable request to the authors ## SUPPLEMENTAL MATERIAL ### S1. Full Search Term for PubMed (“infant, newborn”[MeSH Terms] OR “infan*”[Text Word] OR “neonat*”[Text Word] OR “newborn*”[Text Word] OR “premature”[Text Word] OR “low birth weight”[Text Word] OR “VLBW”[Text Word] OR “LBW”[Text Word] OR “ELBW”[Text Word] OR (“Premature birth”[MeSH Terms] OR “infant, premature”[MeSH Terms] OR “infant, very low birth weight”[MeSH Terms] OR “infant, premature, diseases”[MeSH Terms] OR ((“preterm”[Text Word] OR “premature”[Text Word] OR “Prematurity”[Text Word] OR “prematurely”[Text Word]) AND (“deliver*”[Text Word] OR “birth*”[Text Word] OR “infant*”[Text Word] OR “infancy”[Text Word] OR “baby”[Text Word] OR “babies”[Text Word] OR “neonat*”[Text Word] OR “newborn*”[Text Word] OR “perinatal”[Text Word] OR “parturition”[Text Word])) OR “preemi*”[Text Word] OR “premi”[Text Word] OR “premie”[Text Word] OR “premies”[Text Word] OR “preterm”[Text Word] OR “low birth weight”[Text Word] OR “low birthweight”[Text Word] OR “early gestation*”[Text Word] OR “low gestation*”[Text Word])) AND (“Brain”[MeSH Terms] OR “Brain”[Text Word] OR “nerve fibers, myelinated”[MeSH Terms] OR “white matter*”[Text Word] OR “myelinated nerve fiber*”[Text Word] OR “Glia”[Text Word] OR “gray matter”[Text Word] OR “cerebel*”[Text Word] OR “ventric*”[Text Word] OR “cerebrum”[Text Word] OR “corpus callosum”[Text Word] OR “internal capsules”[Text Word] OR “external capsules”[Text Word] OR “anterior commissure”[Text Word] OR “limbic system”[Text Word]) AND (“hydrocortisone”[MeSH Terms] OR “dexamethasone”[MeSH Terms] OR “betamethasone”[MeSH Terms] OR “steroids”[MeSH Terms] OR “Glucocorticoids”[MeSH Terms] OR “Adrenal Cortex Hormones”[MeSH Terms] OR “hydrocortisone*”[Text Word] OR “betamethason*”[Text Word] OR “dexamethason*”[Text Word] OR “steroid*”[Text Word] OR “corticoster*”[Text Word] OR “corticoid*”[Text Word] OR “glucocortic*”[Text Word] OR “adrenal cortex hormone*”[Text Word]) AND “English”[Language] ### S2. Studies of glucocorticoids that reported a brain injury as a clinical characteristic We identified 50 studies that reported a brain injury outcome, most commonly IVH, as a characteristic of the study population after treatment with corticosteroid, but did not directly assess the effect of the steroid on brain injury. These studies are listed in the **Table**. (Sinkin et al., 2000) is a representative example of this category. Sinkin and colleagues conducted a multi-center, double blind phase 2 randomized controlled trial of a two dose course of dexamethasone early in neonatal life to assess the clinical efficacy and safety of the medication. The study population included infants under 30 weeks gestation with respiratory distress syndrome who required mechanical ventilation at 12-18 hours of life after surfactant. The dosing regimen was 0.5 mg dexamethasone administered via IV at 12-18 hours of life and 12 hours later. Later dexamethasone use was permitted per clinician discretion. The relevant brain outcome was presence and severity of IVH. 384 infants were enrolled and 189 received dexamethasone. There was no difference between the groups in incidence or severity of intraventricular hemorrhage. Of note, LeFlore (LeFlore JL et al., 2002) reported on an investigation of infants who received either no treatment, antenatal dexamethasone, postnatal dexamethasone only, or both antenatal and postnatal steroids. However, the aim of the study and the reported results were on the effect of the antenatal exposure on cranial ultrasound at discharge. They report that the likelihood of abnormal ultrasound at discharge was greater in the postnatal dexamethasone groups compared with the no-postnatal dexamethasone groups. They do not report on the relationship between postnatal dexamethasone and specific cranial ultrasound findings as they do for antenatal dexamethasone at discharge. ### S3. Glucocorticoids as risk factors 12 studies examined perinatal clinical risk factors and their effect on brain development (Batton et al., 2012; Cuzzilla R et al., 2018; Heo et al., 2018; Kelly CE et al., 2014; Kidokoro H et al., 2014; Matthews LG et al., 2018; Neubauer et al., 2017; Parikh NA et al., 2021; Thompson DK et al., 2008; Thompson et al., 2012; Vesoulis ZA et al., 2018; Watterberg et al., 2004). For the most part, steroid type and administration details are not specified. Whether or not steroid administration functions as a proxy for some other factor, such as pulmonary health, was not able to be discerned. View this table: [Table.](http://medrxiv.org/content/early/2022/11/11/2022.11.09.22282133/T3) Table. Studies reporting brain injury as an outcome ## ACKNOWLEDGEMENTS This work was supported by the National Institutes of Health (2RO1-HD069150) and the Young Investigator Award to Dr. Dubner, from the Society of Developmental and Behavioral Pediatrics (2019). * Received November 9, 2022. * Revision received November 9, 2022. * Accepted November 11, 2022. * © 2022, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NonCommercial-NoDerivs 4.0 International), CC BY-NC-ND 4.0, as described at [http://creativecommons.org/licenses/by-nc-nd/4.0/](http://creativecommons.org/licenses/by-nc-nd/4.0/) ## REFERENCES 1. Alison M, Tilea B, Toumazi A, Biran V, Mohamed D, Alberti C, Bourmaud A, Baud O, 2020. Prophylactic hydrocortisone in extremely preterm infants and brain MRI abnormality. Arch Dis Child Fetal Neonatal Ed 105, 520–525. [https://doi.org/10.1136/archdischild-2019-317720](https://doi.org/10.1136/archdischild-2019-317720) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTM6ImZldGFsbmVvbmF0YWwiO3M6NToicmVzaWQiO3M6OToiMTA1LzUvNTIwIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMTEvMTEvMjAyMi4xMS4wOS4yMjI4MjEzMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 2. Arksey, H., O’Malley, L., 2005. Scoping studies: towards a methodological framework. International Journal of Social Research Methodology 8, 19–32. [https://doi.org/10.1080/1364557032000119616](https://doi.org/10.1080/1364557032000119616) 3. Baud, O., 2004. Postnatal steroid treatment and brain development. Arch Dis Child Fetal Neonatal Ed 89, F96–F100. [https://doi.org/10.1136/adc.2003.028696](https://doi.org/10.1136/adc.2003.028696) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTM6ImZldGFsbmVvbmF0YWwiO3M6NToicmVzaWQiO3M6ODoiODkvMi9GOTYiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMi8xMS8xMS8yMDIyLjExLjA5LjIyMjgyMTMzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 4. Benders, M.J.N.L., Groenendaal, F., van Bel, F., Ha Vinh, R., Dubois, J., Lazeyras, F., Warfield, S.K., Hüppi, P.S., de Vries, L.S., 2009. Brain development of the preterm neonate after neonatal hydrocortisone treatment for chronic lung disease. Pediatr. Res. 66, 555–559. [https://doi.org/10.1203/PDR.0b013e3181b3aec5](https://doi.org/10.1203/PDR.0b013e3181b3aec5) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1203/PDR.0b013e3181b3aec5&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19851225&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000271039100013&link_type=ISI) 5. Bohn, M.C., O’Banion, M.K., Young, D.A., Giuliano, R., Hussain, S., Dean, D.O., Cunningham, L.A., 1994. In vitro studies of glucocorticoid effects on neurons and astrocytes. Annals of the New York Academy of Sciences 746, 243–258. [https://doi.org/10.1111/j.1749-6632.1994.tb39241.x](https://doi.org/10.1111/j.1749-6632.1994.tb39241.x) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=7825881&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1994BD11Z00020&link_type=ISI) 6. Chang, Y.P., 2014. Evidence for adverse effect of perinatal glucocorticoid use on the developing brain. Korean J Pediatr 57, 101–109. [https://doi.org/10.3345/kjp.2014.57.3.101](https://doi.org/10.3345/kjp.2014.57.3.101) 7. Committee on Fetus and Newborn, 2002. Postnatal Corticosteroids to Treat or Prevent Chronic Lung Disease in Preterm Infants. Pediatrics 109, 330–338. [https://doi.org/10.1542/peds.109.2.330](https://doi.org/10.1542/peds.109.2.330) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6InBlZGlhdHJpY3MiO3M6NToicmVzaWQiO3M6OToiMTA5LzIvMzMwIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMTEvMTEvMjAyMi4xMS4wOS4yMjI4MjEzMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 8. Cotterrell, M., Balázs, R., Johnson, A.L., 1972. Effects of corticosteroids on the biochemical maturation of rat brain: postnatal cell formation. J Neurochem 19, 2151–2167. [https://doi.org/10.1111/j.1471-4159.1972.tb05124.x](https://doi.org/10.1111/j.1471-4159.1972.tb05124.x) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1471-4159.1972.tb05124.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=5072390&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1972N344600014&link_type=ISI) 9. Covidence systematic review software, n.d. 10. Cummings, J.J., Pramanik, A.K., COMMITTEE ON FETUS AND NEWBORN, 2022. Postnatal Corticosteroids to Prevent or Treat Chronic Lung Disease Following Preterm Birth. Pediatrics e2022057530. [https://doi.org/10.1542/peds.2022-057530](https://doi.org/10.1542/peds.2022-057530) 11. Damsted, S.K., Born, A.P., Paulson, O.B., Uldall, P., 2011. Exogenous glucocorticoids and adverse cerebral effects in children. Eur J Paediatr Neurol 15, 465–477. [https://doi.org/10.1016/j.ejpn.2011.05.002](https://doi.org/10.1016/j.ejpn.2011.05.002) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ejpn.2011.05.002&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21632268&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) 12. De Kloet, E.R., Vreugdenhil, E., Oitzl, M.S., Joëls, M., 1998. Brain corticosteroid receptor balance in health and disease. Endocr Rev 19, 269–301. [https://doi.org/10.1210/edrv.19.3.0331](https://doi.org/10.1210/edrv.19.3.0331) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1210/er.19.3.269&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9626555&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000074008600002&link_type=ISI) 13. Doyle, L.W., 2021. Postnatal Corticosteroids to Prevent or Treat Bronchopulmonary Dysplasia. NEO 118, 244–251. [https://doi.org/10.1159/000515950](https://doi.org/10.1159/000515950) 14. Doyle, L.W., Cheong, J.L.Y., Hay, S., Manley, B.J., Halliday, H.L., 2021a. Late (≥ 7 days) systemic postnatal corticosteroids for prevention of bronchopulmonary dysplasia in preterm infants. Cochrane Database of Systematic Reviews. [https://doi.org/10.1002/14651858.CD001145.pub5](https://doi.org/10.1002/14651858.CD001145.pub5) 15. Doyle, L.W., Cheong, J.L.Y., Hay, S., Manley, B.J., Halliday, H.L., 2021b. Early (< 7 days) systemic postnatal corticosteroids for prevention of bronchopulmonary dysplasia in preterm infants. Cochrane Database of Systematic Reviews. 16. Favrais, G., Tourneux, P., Lopez, E., Durrmeyer, X., Gascoin, G., Ramful, D., Zana-Taieb, E., Baud, O., 2014. Impact of common treatments given in the perinatal period on the developing brain. Neonatology 106, 163–172. [https://doi.org/10.1159/000363492](https://doi.org/10.1159/000363492) 17. Feldman, H.M., Yeatman, J.D., Lee, E.S., Barde, L.H., Gaman-Bean, S., 2010. Diffusion tensor imaging: a review for pediatric researchers and clinicians. Journal of developmental and behavioral pediatrics : JDBP 31, 346–56. [https://doi.org/10.1097/DBP.0b013e3181dcaa8b](https://doi.org/10.1097/DBP.0b013e3181dcaa8b) 18. Gallini, F., Coppola, M., De Rose, D.U., Maggio, L., Arena, R., Romano, V., Cota, F., Ricci, D., Romeo, D.M., Mercuri, E.M., Vento, G., 2021. Neurodevelopmental outcomes in very preterm infants: The role of severity of Bronchopulmonary Dysplasia. Early Human Development 152, 105275. [https://doi.org/10.1016/j.earlhumdev.2020.105275](https://doi.org/10.1016/j.earlhumdev.2020.105275) 19. Greenough, A., 2022. Hydrocortisone to Prevent Bronchopulmonary Dysplasia — Not a Silver Bullet. New England Journal of Medicine 386, 1181–1183. [https://doi.org/10.1056/NEJMe2200247](https://doi.org/10.1056/NEJMe2200247) 20. Gumbinas, M., Oda, M., Huttenlocher, P., 1973. The effects of corticosteroids on myelination of the developing rat brain. Biol Neonate 22, 355–366. [https://doi.org/10.1159/000240568](https://doi.org/10.1159/000240568) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1159/000094851&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=4778314&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1973R713900004&link_type=ISI) 21. Howard, E., Granoff, D.M., 1968. Increased voluntary running and decreased motor coordination in mice after neonatal corticosterone implantation. Exp Neurol 22, 661– 673. [https://doi.org/10.1016/0014-4886(68)90155-6](https://doi.org/10.1016/0014-4886(68)90155-6) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/0014-4886(68)90155-6&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=5709812&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1968C432900012&link_type=ISI) 22. Huang, W.L., Dunlop, S.A., Harper, C.G., 1999. Effect of Exogenous Corticosteroids on the Developing Central Nervous System: A Review. Obstetrical & Gynecological Survey 54, 336–342. 23. Joëls, M., 2007. Role of corticosteroid hormones in the dentate gyrus. Progress in Brain Research 163, 355–370. [https://doi.org/10.1016/S0079-6123(07)63021-0](https://doi.org/10.1016/S0079-6123(07)63021-0) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0079-6123(07)63021-0&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17765729&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000251961200022&link_type=ISI) 24. Joëls, M., 2001. Corticosteroid actions in the hippocampus. Journal of Neuroendocrinology 13, 657–669. [https://doi.org/10.1046/j.1365-2826.2001.00688.x](https://doi.org/10.1046/j.1365-2826.2001.00688.x) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1046/j.1365-2826.2001.00688.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11489082&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000170426500001&link_type=ISI) 25. Johnston, M.V., Ishida, A., Ishida, W.N., Matsushita, H.B., Nishimura, A., Tsuji, M., 2009. Plasticity and injury in the developing brain. Brain and Development 31, 1–10. [https://doi.org/10.1016/j.braindev.2008.03.014](https://doi.org/10.1016/j.braindev.2008.03.014) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.braindev.2008.03.014&link_type=DOI) 26. Kersbergen, K.J., de Vries, L.S., van Kooij, B.J.M., Išgum, I., Rademaker, K.J., van Bel, F., Hüppi, P.S., Dubois, J., Groenendaal, F., Benders, M.J.N.L., 2013. Hydrocortisone treatment for bronchopulmonary dysplasia and brain volumes in preterm infants. J. Pediatr. 163, 666–671.e1. [https://doi.org/10.1016/j.jpeds.2013.04.001](https://doi.org/10.1016/j.jpeds.2013.04.001) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jpeds.2013.04.001&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23706359&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000323985300012&link_type=ISI) 27. Kidokoro, H., Neil, J.J., Inder, T.E., 2013. New MR imaging assessment tool to define brain abnormalities in very preterm infants at term. AJNR Am J Neuroradiol 34, 2208–2214. [https://doi.org/10.3174/ajnr.A3521](https://doi.org/10.3174/ajnr.A3521) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoiYWpuciI7czo1OiJyZXNpZCI7czoxMDoiMzQvMTEvMjIwOCI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIyLzExLzExLzIwMjIuMTEuMDkuMjIyODIxMzMuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 28. Lodygensky, G.A., Rademaker, K., Zimine, S., Gex-Fabry, M., Lieftink, A.F., Lazeyras, F., Groenendaal, F., de Vries, L.S., Huppi, P.S., 2005. Structural and functional brain development after hydrocortisone treatment for neonatal chronic lung disease. Pediatrics 116, 1–7. [https://doi.org/10.1542/peds.2004-1275](https://doi.org/10.1542/peds.2004-1275) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6InBlZGlhdHJpY3MiO3M6NToicmVzaWQiO3M6NzoiMTE2LzEvMSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIyLzExLzExLzIwMjIuMTEuMDkuMjIyODIxMzMuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 29. Mancini, M., Karakuzu, A., Cohen-Adad, J., Cercignani, M., Nichols, T.E., Stikov, N., 2020. An interactive meta-analysis of MRI biomarkers of myelin. Elife 9, e61523. [https://doi.org/10.7554/eLife.61523](https://doi.org/10.7554/eLife.61523) 30. McEwen, B.S., 1994. Corticosteroids and Hippocampal Plasticity. Annals of the New York Academy of Sciences 746, 134–142. [https://doi.org/10.1111/j.1749-6632.1994.tb39223.x](https://doi.org/10.1111/j.1749-6632.1994.tb39223.x) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=7825871&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1994BD11Z00011&link_type=ISI) 31. McEwen, B.S., Angulo, J., Cameron, H., Chao, H.M., Daniels, D., Gannon, M.N., Gould, E., Mendelson, S., Sakai, R., Spencer, R., Woolley, C., 1992. Paradoxical effects of adrenal steroids on the brain: Protection versus degeneration. Biological Psychiatry 31, 177–199. [https://doi.org/10.1016/0006-3223(92)90204-D](https://doi.org/10.1016/0006-3223(92)90204-D) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=1737079&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1992HC27900008&link_type=ISI) 32. Ment, L.R., Hirtz, D., Hüppi, P.S., 2009. Imaging biomarkers of outcome in the developing preterm brain. The Lancet Neurology 8, 1042–1055. [https://doi.org/10.1016/S1474-4422(09)70257-1](https://doi.org/10.1016/S1474-4422(09)70257-1) 33. Murphy BP, Inder TE, Huppi PS, Warfield S, Zientara GP, Kikinis R, Jolesz FA, Volpe JJ, 2001. Impaired cerebral cortical gray matter growth after treatment with dexamethasone for neonatal chronic lung disease. Pediatrics 107, 217–21. [https://doi.org/10.1542/peds.107.2.217](https://doi.org/10.1542/peds.107.2.217) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6InBlZGlhdHJpY3MiO3M6NToicmVzaWQiO3M6OToiMTA3LzIvMjE3IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMTEvMTEvMjAyMi4xMS4wOS4yMjI4MjEzMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 34. Nishi, M., Kawata, M., 2007. Dynamics of glucocorticoid receptor and mineralocorticoid receptor: Implications from live cell imaging studies. Neuroendocrinology 85, 186–192. [https://doi.org/10.1159/000101917](https://doi.org/10.1159/000101917) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1159/000101917&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17446698&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000247150800006&link_type=ISI) 35. Noguchi, K.K., 2014. Glucocorticoid Induced Cerebellar Toxicity in the Developing Neonate: Implications for Glucocorticoid Therapy during Bronchopulmonary Dysplasia. Cells 3, 36–52. [https://doi.org/10.3390/cells3010036](https://doi.org/10.3390/cells3010036) 36. Onland, W., De Jaegere, A.P., Offringa, M., van Kaam, A., 2017. Systemic corticosteroid regimens for prevention of bronchopulmonary dysplasia in preterm infants. Cochrane Database of Systematic Reviews. [https://doi.org/10.1002/14651858.CD010941.pub2](https://doi.org/10.1002/14651858.CD010941.pub2) 37. Parikh, N.A., Kennedy, K.A., Lasky, R.E., McDavid, G.E., Tyson, J.E., 2013. Pilot randomized trial of hydrocortisone in ventilator-dependent extremely preterm infants: effects on regional brain volumes. J. Pediatr. 162, 685–690.e1. [https://doi.org/10.1016/j.jpeds.2012.09.054](https://doi.org/10.1016/j.jpeds.2012.09.054) 38. Parikh NA, Lasky RE, Kennedy KA, Moya FR, Hochhauser L, Romo S, Tyson JE, 2007. Postnatal dexamethasone therapy and cerebral tissue volumes in extremely low birth weight infants. Pediatrics 119, 265–72. [https://doi.org/10.1542/peds.2006-1354](https://doi.org/10.1542/peds.2006-1354) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6InBlZGlhdHJpY3MiO3M6NToicmVzaWQiO3M6OToiMTE5LzIvMjY1IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMTEvMTEvMjAyMi4xMS4wOS4yMjI4MjEzMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 39. Patra, A., Huang, H., Bauer, J.A., Giannone, P.J., 2017. Neurological consequences of systemic inflammation in the premature neonate. Neural Regen Res 12, 890–896. [https://doi.org/10.4103/1673-5374.208547](https://doi.org/10.4103/1673-5374.208547) 40. Rademaker, K.J., de Vries, W.B., 2009. Long-term effects of neonatal hydrocortisone treatment for chronic lung disease on the developing brain and heart. Seminars in Fetal and Neonatal Medicine, Uses and Abuses of Corticosteroids 14, 171–177. [https://doi.org/10.1016/j.siny.2008.11.004](https://doi.org/10.1016/j.siny.2008.11.004) 41. Reul, J.M.H.M., Gesing, A., Droste, S., Stec, I.S.M., Weber, A., Bachmann, C., Bilang-Bleuel, A., Holsboer, F., Linthorst, A.C.E., 2000. The brain mineralocorticoid receptor: Greedy for ligand, mysterious in function. European Journal of Pharmacology 405, 235–249. [https://doi.org/10.1016/S0014-2999(00)00677-4](https://doi.org/10.1016/S0014-2999(00)00677-4) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0014-2999(00)00677-4&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11033331&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000089676800024&link_type=ISI) 42. Rousseau, C., Guichard, M., Saliba, E., Morel, B., Favrais, G., 2021. Duration of mechanical ventilation is more critical for brain growth than postnatal hydrocortisone in extremely preterm infants. Eur J Pediatr 180, 3307–3315. [https://doi.org/10.1007/s00431-021-04113-z](https://doi.org/10.1007/s00431-021-04113-z) 43. Rousseau C, Guichard M, Saliba E, Morel B, Favrais G, 2021. Duration of mechanical ventilation is more critical for brain growth than postnatal hydrocortisone in extremely preterm infants. Eur J Pediatr. [https://doi.org/10.1007/s00431-021-04113-z](https://doi.org/10.1007/s00431-021-04113-z) 44. Shah, S.S., Ohlsson, A., Halliday, H.L., Shah, V.S., 2017. Inhaled versus systemic corticosteroids for preventing bronchopulmonary dysplasia in ventilated very low birth weight preterm neonates. Cochrane Database of Systematic Reviews. [https://doi.org/10.1002/14651858.CD002058.pub3](https://doi.org/10.1002/14651858.CD002058.pub3) 45. Tam, E.W.Y., Chau, V., Ferriero, D.M., Barkovich, A.J., Poskitt, K.J., Studholme, C., Fok, E.D.-Y., Grunau, R.E., Glidden, D.V., Miller, S.P., 2011. Preterm cerebellar growth impairment after postnatal exposure to glucocorticoids. Sci Transl Med 3, 105ra105. [https://doi.org/10.1126/scitranslmed.3002884](https://doi.org/10.1126/scitranslmed.3002884) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6InNjaXRyYW5zbWVkIjtzOjU6InJlc2lkIjtzOjE0OiIzLzEwNS8xMDVyYTEwNSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIyLzExLzExLzIwMjIuMTEuMDkuMjIyODIxMzMuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 46. Volpe, J.J., 2009. Brain injury in premature infants: a complex amalgam of destructive and developmental disturbances. Lancet Neurol 8, 110–24. [https://doi.org/10.1016/S1474-4422(08)70294-1](https://doi.org/10.1016/S1474-4422(08)70294-1) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S1474-4422(08)70294-1&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19081519&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000261860700026&link_type=ISI) 47. Watterberg, K.L., Walsh, M.C., Li, L., Chawla, S., D’Angio, C.T., Goldberg, R.N., Hintz, S.R., Laughon, M.M., Yoder, B.A., Kennedy, K.A., McDavid, G.E., Backstrom-Lacy, C., Das, A., Crawford, M.M., Keszler, M., Sokol, G.M., Poindexter, B.B., Ambalavanan, N., Hibbs, A.M., Truog, W.E., Schmidt, B., Wyckoff, M.H., Khan, A.M., Garg, M., Chess, P.R., Reynolds, A.M., Moallem, M., Bell, E.F., Meyer, L.R., Patel, R.M., Van Meurs, K.P., Cotten, C.M., McGowan, E.C., Hines, A.C., Merhar, S., Peralta-Carcelen, M., Wilson-Costello, D.E., Kilbride, H.W., DeMauro, S.B., Heyne, R.J., Mosquera, R.A., Natarajan, G., Purdy, I.B., Lowe, J.R., Maitre, N.L., Harmon, H.M., Hogden, L.A., Adams-Chapman, I., Winter, S., Malcolm, W.F., Higgins, R.D., 2022. Hydrocortisone to Improve Survival without Bronchopulmonary Dysplasia. N Engl J Med 386, 1121–1131. [https://doi.org/10.1056/NEJMoa2114897](https://doi.org/10.1056/NEJMoa2114897) 48. Zoli, M., Agnati, L.F., Fuxe, K., Ferraguti, F., Biagini, G., Cintra, A., Gustafsson, J.A., 1990. Long-lasting reduction of glucocorticoid receptor immunoreactivity in the hippocampal field CA1 but not in the dentate gyrus after neonatal treatment with corticosterone in the rat. Acta Physiol Scand 138, 577–579. [https://doi.org/10.1111/j.1748-1716.1990.tb08887.x](https://doi.org/10.1111/j.1748-1716.1990.tb08887.x) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=2353583&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) 1. Anttila E, Peltoniemi O, Haumont D, Herting E, ter Horst H, Heinonen K, Kero P, Nykänen P, Oetomo SB, Hallman M, 2005. Early neonatal dexamethasone treatment for prevention of bronchopulmonary dysplasia. Randomised trial and meta-analysis evaluating the duration of dexamethasone therapy. Eur J Pediatr 164, 472–81. [https://doi.org/10.1007/s00431-005-1645-8](https://doi.org/10.1007/s00431-005-1645-8) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s00431-005-1645-8&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15864643&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000230811500002&link_type=ISI) 2. Bassler, D., Plavka, R., Shinwell, E.S., Hallman, M., Jarreau, P.-H., Carnielli, V., Van den Anker, J.N., Meisner, C., Engel, C., Schwab, M., Halliday, H.L., Poets, C.F., 2015. Early Inhaled Budesonide for the Prevention of Bronchopulmonary Dysplasia. N Engl J Med 373, 1497–1506. [https://doi.org/10.1056/NEJMoa1501917](https://doi.org/10.1056/NEJMoa1501917) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1501917&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26465983&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) 3. Batton, B.J., Li, L., Newman, N.S., Das, A., Watterberg, K.L., Yoder, B.A., 2012. Eunice Kennedy Shriver National Institute of Child Health and Human Development Neonatal Research Network. Feasibility study of early blood pressure management in extremely preterm infants. Journal of Pediatrics 161, 65–9. [https://doi.org/10.1016/j.jpeds.2012.01.014](https://doi.org/10.1016/j.jpeds.2012.01.014) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jpeds.2012.01.014&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22336574&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) 4. Baud, O., Maury, L., Lebail, F., Ramful, D., El Moussawi, F., Nicaise, C., Zupan-Simunek, V., Coursol, A., Beuchée, A., Bolot, P., Andrini, P., Mohamed, D., Alberti, C., PREMILOC trial study group, 2016. Effect of early low-dose hydrocortisone on survival without bronchopulmonary dysplasia in extremely preterm infants (PREMILOC): a double-blind, placebo-controlled, multicentre, randomised trial. Lancet 387, 1827–1836. [https://doi.org/10.1016/S0140-6736(16)00202-6](https://doi.org/10.1016/S0140-6736(16)00202-6) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0140-6736(16)00202-6&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26916176&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) 5. Biswas, S., Buffery, J., Enoch, H., Bland, M., Markiewicz, M., Walters, D., 2003. Pulmonary effects of triiodothyronine (T3) and hydrocortisone (HC) supplementation in preterm infants less than 30 weeks’ gestation: results of the THORN trial - thyroid hormone replacement in neonates. Pediatric Research 53, 48–56. [https://doi.org/10.1203/00006450-200301000-00011](https://doi.org/10.1203/00006450-200301000-00011) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1203/01.PDR.0000041512.36915.D4&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12508081&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000180142800010&link_type=ISI) 6. Bonsante, F., Latorre, G., Iacobelli, S., Forziati, V., Laforgia, N., Esposito, L., 2007. Early low-dose hydrocortisone in very preterm infants: a randomized, placebo-controlled trial. Neonatology 91, 217–21. [https://doi.org/10.1159/000098168](https://doi.org/10.1159/000098168) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1159/000098168&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17568152&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000247028900001&link_type=ISI) 7. Bourchier, D., Weston, P.J., 1997. Randomised trial of dopamine compared with hydrocortisone for the treatment of hypotensive very low birthweight infants. Arch Dis Child Fetal Neonatal Ed 76, F174–178. [https://doi.org/10.1136/fn.76.3.f174](https://doi.org/10.1136/fn.76.3.f174) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTM6ImZldGFsbmVvbmF0YWwiO3M6NToicmVzaWQiO3M6OToiNzYvMy9GMTc0IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMTEvMTEvMjAyMi4xMS4wOS4yMjI4MjEzMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 8. Brozanski, B., Jones, J., Gilmour, C., Balsan, M., Vazquez, R., Israel, B., Newman, B., Mimouni, F., Guthrie, R., 1995. Effect of pulse dexamethasone therapy on the incidence and severity of chronic lung disease in the very low birth weight infant. Journal of pediatrics 126, 769–776. [https://doi.org/10.1016/s0022-3476(95)70410-8](https://doi.org/10.1016/s0022-3476(95)70410-8) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0022-3476(95)70410-8&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=7752005&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1995QY15100018&link_type=ISI) 9. Cuzzilla R, Spittle AJ, Lee KJ, Rogerson S, Cowan FM, Doyle LW, Cheong JLY, 2018. Postnatal Brain Growth Assessed by Sequential Cranial Ultrasonography in Infants Born < Weeks’ Gestational Age. AJNR Am J Neuroradiol 39, 1170–1176. [https://doi.org/10.3174/ajnr.A5679](https://doi.org/10.3174/ajnr.A5679) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoiYWpuciI7czo1OiJyZXNpZCI7czo5OiIzOS82LzExNzAiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMi8xMS8xMS8yMDIyLjExLjA5LjIyMjgyMTMzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 10. Doyle, L.W., Davis, P.G., Morley, C.J., McPhee, A., Carlin, J.B., 2006. Low-dose dexamethasone facilitates extubation among chronically ventilator-dependent infants: a multicenter, international, randomized, controlled trial. Pediatrics 117, 75–83. [https://doi.org/10.1542/peds.2004-2843](https://doi.org/10.1542/peds.2004-2843) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6InBlZGlhdHJpY3MiO3M6NToicmVzaWQiO3M6ODoiMTE3LzEvNzUiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMi8xMS8xMS8yMDIyLjExLjA5LjIyMjgyMTMzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 11. Durand, M., Sardesai, S., McEvoy, C., 1995. Effects of early dexamethasone therapy on pulmonary mechanics and chronic lung disease in very low birth weight infants: a randomized, controlled trial. Pediatrics 95, 584–590. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6InBlZGlhdHJpY3MiO3M6NToicmVzaWQiO3M6ODoiOTUvNC81ODQiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMi8xMS8xMS8yMDIyLjExLjA5LjIyMjgyMTMzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 12. Efird, M.M., Heerens, A.T., Gordon, P.V., Bose, C.L., Young, D.A., 2005. A randomized-controlled trial of prophylactic hydrocortisone supplementation for the prevention of hypotension in extremely low birth weight infants. Journal of Perinatology 25, 119–24. [https://doi.org/10.1038/sj.jp.7211193](https://doi.org/10.1038/sj.jp.7211193) 13. Gaissmaier, R.E., Pohlandt, F., 1999. Single-dose dexamethasone treatment of hypotension in preterm infants. Journal of Pediatrics 134, 701–5. [https://doi.org/10.1016/s0022-3476(99)70284-2](https://doi.org/10.1016/s0022-3476(99)70284-2) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0022-3476(99)70284-2&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10356137&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000080820900008&link_type=ISI) 14. Garland, J.S., Alex, C.P., Pauly, T.H., Whitehead, V.L., Brand, J., Winston, J.F., 1999. A three-day course of dexamethasone therapy to prevent chronic lung disease in ventilated neonates: a randomized trial. Pediatrics 104, 91–9. [https://doi.org/10.1542/peds.104.1.91](https://doi.org/10.1542/peds.104.1.91) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6InBlZGlhdHJpY3MiO3M6NToicmVzaWQiO3M6ODoiMTA0LzEvOTEiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMi8xMS8xMS8yMDIyLjExLjA5LjIyMjgyMTMzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 15. Halliday, H.L., Patterson, C.C., Halahakoon, C.W., 2001. A multicenter, randomized open study of early corticosteroid treatment (OSECT) in preterm infants with respiratory illness: comparison of early and late treatment and of dexamethasone and inhaled budesonide. Pediatrics 107, 232–40. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6InBlZGlhdHJpY3MiO3M6NToicmVzaWQiO3M6OToiMTA3LzIvMjMyIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMTEvMTEvMjAyMi4xMS4wOS4yMjI4MjEzMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 16. Heo, J.S., Kim, E.-K., Choi, Y.H., Shin, S.H., Sohn, J.A., Cheon, J.-E., Kim, H.-S., 2018. Timing of sepsis is an important risk factor for white matter abnormality in extremely premature infants with sepsis. Pediatr Neonatol 59, 77–84. [https://doi.org/10.1016/j.pedneo.2017.07.008](https://doi.org/10.1016/j.pedneo.2017.07.008) 17. Kelly CE, Cheong JL, Molloy C, Anderson PJ, Lee KJ, Burnett AC, Connelly A, Doyle LW, Thompson DK, 2014. Neural correlates of impaired vision in adolescents born extremely preterm and/or extremely low birthweight. PLoS One 9, e93188. [https://doi.org/10.1371/journal.pone.0093188](https://doi.org/10.1371/journal.pone.0093188) 18. Kidokoro H, Anderson PJ, Doyle LW, Woodward LJ, Neil JJ, Inder TE, 2014. Brain injury and altered brain growth in preterm infants: predictors and prognosis. Pediatrics 134, e444-53. [https://doi.org/10.1542/peds.2013-2336](https://doi.org/10.1542/peds.2013-2336) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6InBlZGlhdHJpY3MiO3M6NToicmVzaWQiO3M6MTA6IjEzNC8yL2U0NDQiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMi8xMS8xMS8yMDIyLjExLjA5LjIyMjgyMTMzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 19. Kopelman AE, Moise AA, Holbert D, Hegemier SE, 1999. A single very early dexamethasone dose improves respiratory and cardiovascular adaptation in preterm infants. J Pediatr 135, 345–50. [https://doi.org/10.1016/s0022-3476(99)70132-0](https://doi.org/10.1016/s0022-3476(99)70132-0) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0022-3476(99)70132-0&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10484801&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000082662800018&link_type=ISI) 20. Kovács, L., Davis, G., Faucher, D., Papageorgiou, A., 1998. Efficacy of sequential early systemic and inhaled corticosteroid therapy in the prevention of chronic lung disease of prematurity. Acta paediatrica 87, 792–798. [https://doi.org/10.1080/080352598750013905](https://doi.org/10.1080/080352598750013905) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1080/080352598750013905&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9722255&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000075058700016&link_type=ISI) 21. LeFlore JL, Salhab WA, Broyles RS, Engle WD, 2002. Association of antenatal and postnatal dexamethasone exposure with outcomes in extremely low birth weight neonates. Pediatrics 110, 275–9. [https://doi.org/10.1542/peds.110.2.275](https://doi.org/10.1542/peds.110.2.275) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6InBlZGlhdHJpY3MiO3M6NToicmVzaWQiO3M6OToiMTEwLzIvMjc1IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMTEvMTEvMjAyMi4xMS4wOS4yMjI4MjEzMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 22. Lim G, Lee BS, Choi YS, Park HW, Chung ML, Choi HJ, Kim EA, Kim KS, 2015. Delayed Dexamethasone Therapy and Neurodevelopmental Outcomes in Preterm Infants with Bronchopulmonary Dysplasia. Pediatr Neonatol 56, 261–7. [https://doi.org/10.1016/j.pedneo.2014.11.006](https://doi.org/10.1016/j.pedneo.2014.11.006) 23. Lin, Y.J., Yeh, T.F., Hsieh, W.S., Chi, Y.C., Lin, H.C., Lin, C.H., 1999. Prevention of chronic lung disease in preterm infants by early postnatal dexamethasone therapy. Pediatric Pulmonology 27, 21–6. [https://doi.org/10.1002/(sici)1099-0496(199901)27:1](https://doi.org/10.1002/(sici)1099-0496(199901)27:1) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/(SICI)1099-0496(199901)27:1<21::AID-PPUL5>3.0.CO;2-Y&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10023787&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000078272500005&link_type=ISI) 24. Malloy, C.A., Hilal, K., Weiss, M.G., Rizvi, Z., Muraskas, J.K., 2005. A prospective, randomized, double-masked trial comparing low dose to conventional dose dexamethasone in neonatal chronic lung disease. Internet Journal of Pediatrics and Neonatology 5. 25. Matthews LG, Inder TE, Pascoe L, Kapur K, Lee KJ, Monson BB, Doyle LW, Thompson DK, Anderson PJ, 2018. Longitudinal Preterm Cerebellar Volume: Perinatal and Neurodevelopmental Outcome Associations. Cerebellum 17, 610–627. [https://doi.org/10.1007/s12311-018-0946-1](https://doi.org/10.1007/s12311-018-0946-1) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s12311-018-0946-1&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29949094&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) 26. McEvoy, C., Bowling, S., Williamson, K., McGaw, P., Durand, M., 2004. Randomized, double-blinded trial of low-dose dexamethasone: II. Functional residual capacity and pulmonary outcome in very low birth weight infants at risk for bronchopulmonary dysplasia. Pediatric Pulmonology 38, 55–63. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/ppul.20037&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15170874&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000222395100007&link_type=ISI) 27. Mieskonen, S., Eronen, M., Malmberg, L., Turpeinen, M., Kari, M., Hallman, M., 2003. Controlled trial of dexamethasone in neonatal chronic lung disease: an 8-year follow-up of cardiopulmonary function and growth. Acta paediatrica 92, 896–904. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1080/08035250310003451&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12948063&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000184808300004&link_type=ISI) 28. Neubauer, V., Djurdjevic, T., Griesmaier, E., Biermayr, M., Gizewski, E.R., Kiechl-Kohlendorfer, U., 2017. Routine Magnetic Resonance Imaging at Term-Equivalent Age Detects Brain Injury in 25% of a Contemporary Cohort of Very Preterm Infants. PLoS ONE 12, e0169442. [https://doi.org/10.1371/journal.pone.0169442](https://doi.org/10.1371/journal.pone.0169442) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0169442&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28046071&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) 29. Ng, P.C., Lee, C.H., Bnur, F.L., Chan, I.H., Lee, A.W., Wong, E., 2006. A double-blind, randomized, controlled study of a “stress dose” of hydrocortisone for rescue treatment of refractory hypotension in preterm infants. Pediatrics 117, 367–75. [https://doi.org/10.1542/peds.2005-0869](https://doi.org/10.1542/peds.2005-0869) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6InBlZGlhdHJpY3MiO3M6NToicmVzaWQiO3M6OToiMTE3LzIvMzY3IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMTEvMTEvMjAyMi4xMS4wOS4yMjI4MjEzMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 30. Odd, D.E., Armstrong, D.L., Teele, R.L., Kuschel, C.A., Harding, J.E., 2004. A randomized trial of two dexamethasone regimens to reduce side-effects in infants treated for chronic lung disease of prematurity. Journal of Paediatrics and Child Health 40, 282–9. [https://doi.org/10.1111/j.1440-1754.2004.00364.x](https://doi.org/10.1111/j.1440-1754.2004.00364.x) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1440-1754.2004.00364.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15151582&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000221437500008&link_type=ISI) 31. Onland, W., Cools, F., Kroon, A., Rademaker, K., Merkus, M.P., Dijk, P.H., 2019. Effect of hydrocortisone therapy initiated 7 to 14 days after birth on mortality or bronchopulmonary dysplasia among very preterm infants receiving mechanical ventilation: a randomized clinical trial. JAMA 321, 354–63. [https://doi.org/10.1001/jama.2018.21443](https://doi.org/10.1001/jama.2018.21443) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) 32. Parikh, N., Kennedy, K., Lasky, R., Tyson, J., 2015. Neurodevelopmental Outcomes of Extremely Preterm Infants Randomized to Stress Dose Hydrocortisone. PloS one 10, e0137051. [https://doi.org/10.1371/journal.pone.0137051](https://doi.org/10.1371/journal.pone.0137051) 33. Parikh NA, Sharma P, He L, Li H, Altaye M, Priyanka Illapani VS, 2021. Perinatal Risk and Protective Factors in the Development of Diffuse White Matter Abnormality on Term-Equivalent Age Magnetic Resonance Imaging in Infants Born Very Preterm. J Pediatr 233, 58–65.e3. [https://doi.org/10.1016/j.jpeds.2020.11.058](https://doi.org/10.1016/j.jpeds.2020.11.058) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jpeds.2020.11.058&link_type=DOI) 34. Peltoniemi, O., Kari, M.A., Heinonen, K., Saarela, T., Nikolajev, K., Andersson, S., 2005. Pretreatment cortisol values may predict responses to hydrocortisone administration for the prevention of bronchopulmonary dysplasia in high-risk infants. Journal of Pediatrics 146, 632–7. [https://doi.org/10.1016/j.jpeds.2004.12.040](https://doi.org/10.1016/j.jpeds.2004.12.040) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jpeds.2004.12.040&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15870666&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000229109000012&link_type=ISI) 35. Rastogi, A., Akintorin, S.M., Bez, M.L., Morales, P., Pildes, R.S., 1996. A controlled trial of dexamethasone to prevent bronchopulmonary dysplasia in surfactant-treated infants. Pediatrics 98, 204–10. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6InBlZGlhdHJpY3MiO3M6NToicmVzaWQiO3M6ODoiOTgvMi8yMDQiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMi8xMS8xMS8yMDIyLjExLjA5LjIyMjgyMTMzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 36. Romagnoli, C., Zecca, E., Luciano, R., Torrioli, G., Tortorolo, G., 2002. Controlled trial of early dexamethasone treatment for the prevention of chronic lung disease in preterm infants: a 3-year follow-up. Pediatrics 109. [https://doi.org/10.1542/peds.109.6.e85](https://doi.org/10.1542/peds.109.6.e85) 37. Romagnoli, C, Zecca, E., Luciano, R., Torrioli, G., Tortorolo, G., 2002. A three year follow-up of preterm infants after moderately early treatment with dexamethasone. Archives of disease of childhood fetal and neonatal edition 87, F55–8. 38. Romagnoli C, Zecca E, Vento G, De Carolis MP, Papacci P, Tortorolo G, 1999. Early postnatal dexamethasone for the prevention of chronic lung disease in high-risk preterm infants. Intensive Care Med 25, 717–21. [https://doi.org/10.1007/s001340050935](https://doi.org/10.1007/s001340050935) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s001340050935&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10470576&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000081856900012&link_type=ISI) 39. Sanders, R.J., Cox, C., Phelps, D.L., Sinkin, R.A., 1994. Two doses of early intravenous dexamethasone for the prevention of bronchopulmonary dysplasia in babies with respiratory distress syndrome. Pediatric Research 36, 122–8. [https://doi.org/10.1203/00006450-199407001-00022](https://doi.org/10.1203/00006450-199407001-00022) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1203/00006450-199407001-00022&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=7936832&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1994NT46200020&link_type=ISI) 40. Shinwell, E., Karplus, M., Reich, D., 1999. Early dexamethasone therapy is associated with increased incidence of cerebral palsy 240–254. 41. Shinwell, E.S., Karplus, M., Zmora, E., Reich, D., Rothschild, A., Blazer, S., 1996. Failure of early postnatal dexamethasone to prevent chronic lung disease in infants with respiratory distress syndrome. Archives of Disease in Childhood. Fetal and Neonatal Edition 74. [https://doi.org/10.1136/fn.74.1.f33](https://doi.org/10.1136/fn.74.1.f33) 42. Sinkin, R.A., Dweck, H.S., Horgan, M.J., Gallaher, K.J., Cox, C., Maniscalco, W.M., 2000. Early dexamethasone - attempting to prevent chronic lung disease. Pediatrics 105, 542–8. [https://doi.org/10.1542/peds.105.3.542](https://doi.org/10.1542/peds.105.3.542) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6InBlZGlhdHJpY3MiO3M6NToicmVzaWQiO3M6OToiMTA1LzMvNTQyIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMTEvMTEvMjAyMi4xMS4wOS4yMjI4MjEzMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 43. Smets, K., Schwagten, B., 2000. Postnatal cystic germinolysis and neonatal chronic lung disease: evaluation of risk factors and neurodevelopmental outcome. Acta Paediatr 89, 1111– 1114. [https://doi.org/10.1080/713794556](https://doi.org/10.1080/713794556) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1080/713794556&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11071094&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000089849800018&link_type=ISI) 44. Smolkin, T., Ulanovsky, I., Jubran, H., Blazer, S., Makhoul, I.R., 2014. Experience with oral betamethasone in extremely low birthweight infants with bronchopulmonary dysplasia. Archives of Disease in Childhood. Fetal and Neonatal Edition 99. [https://doi.org/10.1136/archdischild-2014-306619](https://doi.org/10.1136/archdischild-2014-306619) 45. Soll, R.F., 1999. Vermont Oxford Network Steroid Study Group. Early postnatal dexamethasone therapy for the prevention of chronic lung disease. Pediatric Research 45. 46. Stark, A.R., Carlo, W.A., Tyson, J.E., Papile, L.A., Wright, L.L., Shankaran, S., 2001. National Institute of Child Health and Human Development Neonatal Research Network. Adverse effects of early dexamethasone in extremely-low-birth-weight infants. New England Journal of Medicine 344, 95–101. [https://doi.org/10.1056/NEJM200101113440203](https://doi.org/10.1056/NEJM200101113440203) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJM200101113440203&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11150359&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000166281100003&link_type=ISI) 47. Subhedar, N.V., Ryan, S.W., Shaw, N.J., 1997. Open randomised controlled trial of inhaled nitric oxide and early dexamethasone in high risk preterm infants. Archives of Disease in Childhood. Fetal and Neonatal Edition 77. [https://doi.org/10.1136/fn.77.3.f185](https://doi.org/10.1136/fn.77.3.f185) 48. Suske, G., Oestreich, K., Varnholt, V., Lasch, P., Kachel, W., 1996. Influence of early postnatal dexamethasone therapy on ventilator dependency in surfactant-substituted preterm infants. Acta Paediatrica 85, 713–8. [https://doi.org/10.1111/j.1651-2227.1996.tb14132.x](https://doi.org/10.1111/j.1651-2227.1996.tb14132.x) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1651-2227.1996.tb14132.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=8816210&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1996UV57900016&link_type=ISI) 49. Takayanagi, T., Matsuo, K., Egashira, T., Mizukami, T., 2015. Neonatal hydrocortisone therapy does not have a serious suppressive effect on the later function of the hypothalamus-pituitary-adrenal axis. Acta Paediatr. 104, e195–199. [https://doi.org/10.1111/apa.12926](https://doi.org/10.1111/apa.12926) 50. Tapia, J.L., Ramirez, R., Cifuentes, J., Fabres, J., Hubner, M.E., Bancalari, A., 1998. The effect of early dexamethasone administration on bronchopulmonary dysplasia in preterm infants with respiratory distress syndrome. Journal of Pediatrics 132, 48–52. [https://doi.org/10.1016/s0022-3476(98)70483-4](https://doi.org/10.1016/s0022-3476(98)70483-4) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0022-3476(98)70483-4&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9469999&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000071796700010&link_type=ISI) 51. Thompson, D.K., Inder, T.E., Faggian, N., Warfield, S.K., Anderson, P.J., Doyle, L.W., Egan, G.F., 2012. Corpus callosum alterations in very preterm infants: perinatal correlates and 2 year neurodevelopmental outcomes. Neuroimage 59, 3571–81. [https://doi.org/10.1016/j.neuroimage.2011.11.057](https://doi.org/10.1016/j.neuroimage.2011.11.057) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.neuroimage.2011.11.057&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22154956&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000301090100053&link_type=ISI) 52. Thompson DK, Wood SJ, Doyle LW, Warfield SK, Lodygensky GA, Anderson PJ, Egan GF, Inder TE, 2008. Neonate hippocampal volumes: prematurity, perinatal predictors, and 2-year outcome. Ann Neurol 63, 642–51. [https://doi.org/10.1002/ana.21367](https://doi.org/10.1002/ana.21367) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/ana.21367&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18384167&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000255960600013&link_type=ISI) 53. Tsukahara, H., Watanabe, Y., Yasutomi, M., Kobata, R., Tamura, S., Kimura, K., 1999. Early (4-7 days of age) dexamethasone therapy for prevention of chronic lung disease in preterm infants. Biology of the Neonate 76, 283–90. [https://doi.org/10.1159/000014170](https://doi.org/10.1159/000014170) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1159/000014170&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10516395&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000083104300004&link_type=ISI) 54. Verma RP, Dasnadi S, Zhao Y, Chen HH, 2017. A comparative analysis of ante- and postnatal clinical characteristics of extremely premature neonates suffering from refractory and non-refractory hypotension: Is early clinical differentiation possible? Early Hum Dev 113, 49–54. [https://doi.org/10.1016/j.earlhumdev.2017.07.010](https://doi.org/10.1016/j.earlhumdev.2017.07.010) 55. Vesoulis ZA, Herco M, Mathur AM, 2018. Divergent risk factors for cerebellar and intraventricular hemorrhage. J Perinatol 38, 278–284. [https://doi.org/10.1038/s41372-017-0010-x](https://doi.org/10.1038/s41372-017-0010-x) 56. Watterberg, K.L., Gerdes, J.S., Cole, C.H., Aucott, S.W., Thilo, E.H., Mammel, M.C., 2004. Prophylaxis of early adrenal insufficiency to prevent bronchopulmonary dysplasia: a multicenter trial. Pediatrics 114, 1649–57. [https://doi.org/10.1542/peds.2004-1159](https://doi.org/10.1542/peds.2004-1159) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6InBlZGlhdHJpY3MiO3M6NToicmVzaWQiO3M6MTA6IjExNC82LzE2NDkiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMi8xMS8xMS8yMDIyLjExLjA5LjIyMjgyMTMzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 57. Watterberg, K.L., Shaffer, M.L., Mishefske, M.J., Leach, C.L., Mammel, M.C., Couser, R.J., Abbasi, S., Cole, C.H., Aucott, S.W., Thilo, E.H., Rozycki, H.J., Lacy, C.B., 2007. Growth and neurodevelopmental outcomes after early low-dose hydrocortisone treatment in extremely low birth weight infants. Pediatrics 120, 40–48. [https://doi.org/10.1542/peds.2006-3158](https://doi.org/10.1542/peds.2006-3158) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6InBlZGlhdHJpY3MiO3M6NToicmVzaWQiO3M6ODoiMTIwLzEvNDAiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMi8xMS8xMS8yMDIyLjExLjA5LjIyMjgyMTMzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 58. Yaseen, H., Okash, I., Hanif, M., al-Umran, K., al-Faraidy, A., 1999. Early dexamethasone treatment in preterm infants treated with surfactant: a double blind controlled trial. Journal of Tropical Pediatrics 45, 304–6. [https://doi.org/10.1093/tropej/45.5.304](https://doi.org/10.1093/tropej/45.5.304) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/tropej/45.5.304&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10584476&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) 59. Yates, H., Chiocchia, V., Linsell, L., Orsi, N., Juszczak, E., Johnson, K., 2019. Very low-dose dexamethasone to facilitate extubation of preterm babies at risk of bronchopulmonary dysplasia: the MINIDEX feasibility RCT. Efficacy and Mechanism Evaluation 6. [https://doi.org/10.3310/eme06080](https://doi.org/10.3310/eme06080) 60. Yeh, T.F., Lin, Y.J., Hsieh, W.S., Lin, H.C., Lin, C.H., Chen, J.Y., 1997. Early postnatal dexamethasone therapy for the prevention of chronic lung disease in preterm infants with respiratory distress syndrome: a multicenter clinical trial. Pediatrics 100. [https://doi.org/10.1542/peds.100.4.e3](https://doi.org/10.1542/peds.100.4.e3) 61. Yeh, T.F., Lin, Y.J., Huang, C.C., Chen, Y.J., Lin, C.H., Lin, H.C., 1998. Early dexamethasone therapy in preterm infants: a follow-up study. Pediatrics 101. [https://doi.org/10.1542/peds.101.5.e7](https://doi.org/10.1542/peds.101.5.e7) 62. Yeh, T.F., Torre, J.A., Rastogi, A., Anyebuno, M.A., Pildes, R.S., 1990. Early postnatal dexamethasone therapy in premature infants with severe respiratory distress syndrome: a double-blind, controlled study. Journal of Pediatrics 117, 273–82. [https://doi.org/10.1016/s0022-3476(05)80547-5](https://doi.org/10.1016/s0022-3476(05)80547-5) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0022-3476(05)80547-5&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=2199642&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F11%2F2022.11.09.22282133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1990DU14800022&link_type=ISI)